throbber
Glatiramer acetate (Copaxone®) induces degenerate,
`Th2-polarized immune responses in patients
`with multiple sclerosis
`
`Petra W. Duda, Mascha C. Schmied, Sandra L. Cook, Jeffrey I. Krieger,
`and David A. Hafler
`
`Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and
`Harvard Medical School, Boston, Massachusetts 02115, USA
`Petra W. Duda’s present address is: Neurology and Experimental Immunology, Department of Research,
`University Hospital Basel, 4031 Basel, Switzerland.
`
`Address correspondence to: David A. Hafler, Center for Neurologic Diseases, Harvard Institutes of Medicine,
`Room 780, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA.
`Phone: (617) 525-5330; Fax: (617) 525-5333; E-mail: hafler@cnd.bwh.harvard.edu.
`
`Petra W. Duda and Mascha C. Schmied contributed equally to this work.
`
`Received for publication November 23, 1999, and accepted in revised form February 15, 2000.
`
`We examined the effect of glatiramer acetate, a random copolymer of alanine, lysine, glutamic acid,
`and tyrosine, on antigen-specific T-cell responses in patients with multiple sclerosis (MS). Glatiramer
`acetate (Copaxone) functioned as a universal antigen, inducing proliferation, independent of any
`prior exposure to the polymer, in T-cell lines prepared from MS or healthy subjects. However, for most
`patients, daily injections of glatiramer acetate abolished this T-cell response and promoted the secre-
`tion of IL-5 and IL-13, which are characteristic of Th2 cells. The surviving glatiramer acetate–reac-
`tive T cells exhibited a greater degree of degeneracy as measured by cross-reactive responses to com-
`binatorial peptide libraries. Thus, it appears that, in some individuals, in vivo administration of
`glatiramer acetate induces highly cross-reactive T cells that secrete Th2 cytokines. To our knowledge,
`glatiramer acetate is the first agent that suppresses human autoimmune disease and alters immune
`function by engaging the T-cell receptor. This compound may be useful in a variety of autoimmune
`disorders in which immune deviation to a Th2 type of response is desirable.
`J. Clin. Invest. 105:967–976 (2000).
`
`Introduction
`Multiple sclerosis (MS) is an inflammatory disease of
`the central nervous system (CNS) white matter. The
`high frequency of activated, myelin-reactive T cells in
`the circulation and cerebrospinal fluid of patients
`with MS is consistent with the hypothesis that an ini-
`tiating event linked to an antecedent microbial infec-
`tion in a genetically susceptible host eventually leads
`to an autoimmune-mediated destruction of myelin
`followed by the surrounding axons (1). After the initi-
`ating event(s), the CNS itself may become a potential
`depot of antigen and MHC, with expression of critical
`second signals required for T-cell activation such as
`B7-1 and CD40 (2, 3) leading to epitope spreading (4).
`MS is thought to be a Th1-mediated disease based
`largely on pathological resemblance to a delayed-type
`hypersensitivity response in the CNS and from obser-
`vations made in the murine experimental autoim-
`mune encephelomyelitis (EAE) model. However, direct
`cloning of myelin-reactive T cells from the blood of
`patients with MS suggests that the majority of T cells
`can secrete both Th1- and Th2-type cytokines (5).
`A major goal in the treatment of autoimmune dis-
`eases has been the development of antigen-specific
`
`therapies that target autoreactive T cells. The discovery
`of epitope spreading in the EAE model (4, 6) and obser-
`vations of diverse T-cell receptor repertoires in response
`to self-antigens have theoretically made this approach
`less attractive. Instead, the concept of bystander sup-
`pression has emerged in which autoreactive Th2 or Th3
`T cells are generated that migrate to the inflamed tar-
`get organ where they are antigen specifically reactivat-
`ed, leading to the secretion of cytokines that downreg-
`ulate inflammation in the local milieu in an antigen
`nonspecific mechanism (7). Two approaches have
`emerged for inducing immune deviation of autoreac-
`tive T cells: mucosal administration of antigen, which
`induces Th2 T-cell responses to the antigen (7), and
`altered peptide ligands (APLs), which, by inducing a
`weaker strength of signal, lead to Th2 deviation of
`cytokine secretion (8–11). Both approaches have been
`used in clinical trials to treat patients with MS, but to
`date, without success.
`An alternative approach to the use of a single self-
`antigen that has been altered or given mucosally is the
`administration of peptide mixtures that contain many
`different antigen specificities. The use of random co-
`polymers that contain amino acids commonly used as
`
`The Journal of Clinical Investigation | April 2000 | Volume 105 | Number 7
`
`967
`
`Page 1 of 10
`
`YEDA EXHIBIT NO. 2069
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`

`mechanism of action of GA involved the induction of
`regulatory T cells. Later, adoptive transfer of GA-spe-
`cific T cells was found to inhibit EAE (22). It was orig-
`inally thought that GA was structurally cross-reactive
`with MBP, although this has remained controversial.
`Recently, TCR antagonism has been suggested to
`occur in addition to competition for MHC binding
`(23). Stimulation of murine GA-reactive T-cell lines
`and clones with MBP was reported to induce the secre-
`tion of Th2 and Th3 cytokines to this cross-reactive
`antigen (24).
`Taken together, these data led to the hypothesis that
`GA acts as an APL in vivo, leading to alterations of
`responses to myelin antigens by cytokine deviation of
`myelin-specific T cells and bystander suppression
`mediated by GA-reactive T cells. Here, we directly test-
`ed this hypothesis by investigating changes in antigen-
`specific responses in patients with MS who were under-
`going treatment with daily subcutaneous injections of
`GA. T-cell reactivity to GA, the immunodominant MBP
`epitope 84–102 as a model myelin antigen, combinato-
`rial libraries derived from the MBP 84-102 sequence,
`and a completely random 13mer sequence was exam-
`ined in vitro before and during a year of treatment.
`Examination of proliferative responses to combinato-
`rial libraries was deemed potentially informative based
`on the observation that combinatorial peptide libraries
`are a powerful tool to examine the degree of T-cell
`receptor degeneracy. That is, the degree to which a T-
`cell clone proliferates to a random combinatorial pep-
`tide library where all of the 13 amino acids are random,
`representing a total of 1913independent peptides, to a
`first approximation provides information regarding
`the degree of degeneracy for that clone, i.e., the more
`peptides the clone can recognize, the more degenerate
`the T-cell receptor. Together, these experiments
`enabled us to examine whether daily subcutaneous
`injections of GA induced alterations of the T-cell
`immune response.
`
`Methods
`Patients. Patients with RR MS in early stages of the dis-
`ease, with MRI findings consistent with the diagnosis,
`and who decided with their physicians to initiate GA
`treatment participated in the trial. No clinical exami-
`
`Table 1
`Characteristics of the study patients
`
`MHC anchors and T-cell receptor (TCR) contact
`residues are possible “universal APLs.” Glatiramer
`acetate (GA) (Copaxone; Teva Marion Partners, Kansas
`City, Missouri, USA) (12) is a random sequence
`polypeptide of the 4 amino acids alanine (A), lysine (K),
`glutamate (E), and tyrosine (Y) at a molar ratio of
`A/K/E/Y of 4.5:3.6:1.5:1, respectively, and an average
`length of 40–100 amino acids. Directly labeled GA effi-
`ciently binds to different murine H-2 I-A molecules and
`to the human counterparts, MHC class II DR, but not
`to DQ or MHC class I, molecules in vitro (13). Bio-
`chemical studies revealed that GA also binds directly
`and with high affinity to purified HLA-DR1, -DR2, and
`-DR4 (14), suggesting that GA contains multiple epi-
`topes enabling it to bind promiscuously to MHC class
`II molecules, where it could potentially be recognized
`by CD4 T cells.
`A “universal antigen” containing multiple epitopes
`would be expected to induce proliferation in vitro, as
`measured by [3H]thymidine incorporation in naive T
`cells from the circulation, representing a high degree
`of cross-reactivity to other peptide antigens. In in vitro
`cultures of PBMCs from healthy humans, a strong
`dose-dependent proliferative response to GA has been
`reported (15). Similarly in our own studies, we found
`that GA elicits dose-dependent responses in all of
`more than 50 humans, including healthy subjects and
`patients with relapsing remitting (RR) and chronic
`progressive MS (P.W. Duda and D.A. Hafler, manu-
`script in preparation). The response to GA could be
`blocked by anti-DR antibodies and the restriction of
`GA-reactive CD4+ T cells to a particular HLA DR mol-
`ecule could be shown on a clonal level. The high pro-
`liferative and cytokine responses of naive PBMC CD4+
`T cells suggest a high frequency of circulating GA-reac-
`tive precursor T cells. Our own limiting dilution analy-
`sis suggests that the precursor frequency of GA-reac-
`tive T cells ranges from 1:5,000 to 1:100,000 PBMCs.
`Thus, GA appears to constitute a highly cross-reactive
`antigen preparation.
`In animal models of MS, prophylactic subcutaneous
`administration of GA has been shown to prevent EAE
`induced by injection of purified myelin basic protein
`(MBP) (12), proteolipid protein (PLP) (16), or myelin
`oligodendrocyte glycoprotein (MOG) (17). Of greater
`importance, in a phase III clinical trial
`subcutaneous administration of GA has
`been shown to decrease the rate of exac-
`erbations and to decrease the appear-
`ance of new lesions, based on magnetic
`resonance imaging (MRI), of patients
`with RR MS (18–20). This represents
`perhaps the first successful use of an
`agent that ameliorates autoimmune dis-
`ease by altering signals presumably
`through the TCR.
`The early observation that cyclophos-
`phamide abrogated the beneficial effect
`of GA on EAE (21), suggested that the
`
`Patient
`1
`2
`3
`4
`5
`6
`7
`
`Sex
`F
`M
`F
`F
`M
`F
`F
`
`DR
`
`3/13
`2/4
`2/4
`3/4
`1/11
`2/4
`2/8
`
`Age at
`onset of disease
`42
`48
`36
`35
`28
`45
`24
`
`Duration
`of disease (y)
`7
`10
`2
`1
`2
`1
`2
`
`EDSSA
`
`AttacksB
`
`1
`3
`1
`2
`1
`1
`0
`
`4
`0
`3
`1
`2
`2
`0
`
`968
`
`The Journal of Clinical Investigation | April 2000 | Volume 105 | Number 7
`
`AAssessed before initiation of treatment. BNumber of attacks in the 2 years before initiation of treatment.
`
`Page 2 of 10
`
`YEDA EXHIBIT NO. 2069
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`

`Figure 1
`The proliferative response to GA is
`decreased on average after daily injec-
`tions of GA. The antigen-specific prolif-
`erative response of 20 or 30 primary T-
`cell lines induced with 40 m g/mL GA as
`described in Methods was measured by
`split-well assay for each patient at each
`time point. Before and at 6 months of
`treatment, all 7 patients could be tested;
`at 3 and 12 months, data for 5 patients
`were obtained. (a) Each panel represents
`data from an individual patient. Squares
`represent mean ± SEM proliferation in D
`cpm of the GA-specific response com-
`pared with the no-antigen control. Back-
`ground levels of the [3H]thymidine incor-
`poration for all patients of the no-antigen
`condition were 1,747 ± 111 before treat-
`ment, and 3,286 ± 175, 3,785 ± 262,
`and 3,509 ± 239 at 3, 6, and 12 months
`of treatment, respectively. The numbers
`in the figure indicate the SI over the no-
`antigen control for each time point. (b)
`The mean stimulation index SI ± SEM for
`all T-cell lines from all patients tested at
`each time point is shown.
`
`nation other than routine follow-up in the clinic was
`performed, and no other preselection criteria were
`applied. Informed consent was obtained before enroll-
`ment, and the study was performed in compliance with
`the rules of the ethical guidelines for human experi-
`ments of the Institutional Review Board of the
`Brigham and Women’s Hospital. Table 1 summarizes
`the patient characteristics.
`Antigens. GA (Copaxone; lots 123211 and 123243) was
`supplied by Teva Marion Partners. MBP 84-102 (DEN-
`PVVHFFKNIVTPRTPP) and MBP 93R (ENPVVHFFRNIVT-
`PR) peptides were synthesized by standard fmoc tech-
`nology and HPLC-purified to greater than 99%.
`Combinatorial peptide library X13 was a 13mer ran-
`domized at each position, and combinatorial peptide
`libraries with random amino acids inserted at position
`X of the MBP 85-99 peptide (ENPVVHFFKNIVTPR) were:
`90X (ENPVVXFFKNIVTPR), 91X (ENPVVHXFKNIVTPR),
`93X (ENPVVHFFXNIVTPR), 90X93R (ENPVVXFFRNIVT-
`PR), and 91X93R (ENPVVHXFRNIVTPR). All peptide
`libraries were obtained from Chiron Technologies
`(Raleigh, North Carolina, USA). Peptides were dissolved
`at 10 mg/mL in DMSO.
`Generation of antigen-specific T-cell lines. PBMCs were
`isolated from fresh drawn heparinized blood by
`Ficoll-Paque (Amersham Pharmacia Biotech, Uppsala,
`Sweden) gradient centrifugation according to manu-
`facturer’s protocol. Antigen-specific T-cell lines were
`generated by culturing 150,000 PBMCs per well in the
`presence of 40 m g/mL GA or MBP 84-102 peptide.
`Unless otherwise indicated, all cell cultures were done
`in 96-well U-bottom microtiter plates in 200 m L com-
`
`plete medium (RPMI 1640; BioWhittaker Inc., Walk-
`ersville, Maryland, USA) containing 2.5% heat-inacti-
`vated pooled human AB serum (PelFreeze, Brown
`Deer, Wisconsin, USA), sodium pyruvate, HEPES,
`nonessential amino acids, and glutamine) in a humid-
`ified 8% CO2 incubator at 37°C. For patient 5, whole
`human MBP was used instead of the MBP 84-102
`peptide throughout. On day 5 of culture, 120 m L of
`culture supernatants was removed and replaced by
`140 m L complete medium containing 10% phyto-
`hemagglutinin-free (PHA-free) T-stim (Collaborative
`Biomedical Laboratories, Bedford, Massachusetts,
`USA). On day 7, each GA-induced T-cell line was
`transferred into 1 mL of complete medium contain-
`ing 10% T-stim for further expansion. On day 12,
`split-well assays were performed to test for antigen-
`specific and cross-reactive proliferation and cytokine
`secretion. For patients 1, 2, 3, and 4, 40 T-cells lines to
`MBP 84-102 and 20 T-cell lines to GA were generated
`at each time point. For patients 5, 6, and 7, 30 T-cell
`lines were generated for each antigen.
`Cross-reactivity assays. Equal aliquots of primary T-cell
`lines were stimulated with irradiated (33 Gy) autolo-
`gous PBMCs that had been preincubated with antigen
`in 96-well U-bottom microtiter and ELISPOT plates at
`37°C for 1 hour. The conditions tested with primary
`GA-reactive lines were 20 m g/mL GA, 100 m g/mL X13,
`40 m g/mL MBP 84-102, 20 m g/mL 90X, 20 m g/mL 91X,
`20 m g/mL 93X, 20 m g/mL 93R, 20 m g/mL 90X93R, 20
`m g/mL 91X93R, and the no-antigen control. Primary
`MBP 84-102–induced lines were tested with 20 m g/mL
`MBP 84-102, 40 m g/mL GA, and no antigen.
`
`The Journal of Clinical Investigation | April 2000 | Volume 105 | Number 7
`
`969
`
`Page 3 of 10
`
`YEDA EXHIBIT NO. 2069
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`

`Proliferation assay and cytokine measurement by ELISA.
`Equal aliquots of primary T-cell lines were stimulated
`with antigen-pulsed autologous PBMCs (100,000 per
`well). After 48 hours, 160 m L of supernatant was
`removed and frozen at –80°C for future cytokine analy-
`sis. The cells were pulsed with 1 m Ci/well of [3H]thymi-
`dine in 100 m L of complete medium. After a further 24
`hours, cells were harvested onto filter paper, and incor-
`poration of [3H]thymidine was measured in a scintilla-
`tion counter (Wallace, Gaithersburg, Maryland, USA).
`Supernatants were tested for cytokines in duplicate by
`performing standard sandwich ELISA using matched
`antibody pairs according to the manufacturer’s protocol
`(Endogen Inc., Woburn, Massachusetts, USA).
`ELISPOT assay. ELISPOT plates (Millipore Corp., Bed-
`ford, Massachusetts, USA) were coated with an optimal
`concentration of 100 m L of primary antibody diluted in
`0.1 mM NaHCO3 (pH 8.3) and incubated overnight at
`4°C. Antibody pairs were the same as those used in the
`sandwich ELISA assay described earlier here. Plates
`were washed 3 times with PBS and blocked with 1%
`BSA in HBSS at 37°C for 1 hour. Plates were again
`washed with PBS 3 times, and antigen-presenting cells
`were added together with antigen and placed in a 37°C,
`8% CO2 incubator for 60 minutes. Responder T cells
`were added, and plates were placed in a 37°C incubator
`for 24 hours. The plates were then washed 3 times with
`TP buffer (0.05% Tween in PBS) and incubated with
`100 m L of biotinylated secondary antibody in TP buffer
`overnight at 4°C. Plates were washed again 3 times
`with TP buffer and incubated at room temperature
`with 100 m L of a 1:1,000 dilution of streptavidin alka-
`line phosphatase conjugate Extravidin (Sigma Chemi-
`cal Co., St. Louis, Missouri, USA) for 2 hours. Plates
`
`were washed, and viewing of spots was carried out with
`100 m L BCIP/NBT substrate (Sigma Chemical Co.) pre-
`pared according to manufacturer’s instructions and by
`developing in dark for up to 20 minutes. The reaction
`was stopped by washing the plates with distilled water.
`Measurement of T-cell proliferation in primary in vitro cul-
`ture. PBMCs, isolated from heparinized blood as already
`described here, were incubated at 50,000 PBMCs per
`well in the presence of PHA or at 150,000 PBMCs per
`well in the presence of tetanus toxoid (Massachusetts
`Biological Laboratories, Jamaica Plain, Massachusetts,
`USA). On day 6, 160 m L of culture supernatant was
`removed from each well and replaced by 100 m L of com-
`plete medium containing 1 m Ci of [3H]thymidine. After
`further incubation at 37°C for 18 hours, cells were har-
`vested onto filter paper, and thymidine incorporation
`was measured by scintillation counting.
`HLA typing. Determination of the HLA DR and DQ
`phenotypes of each patient was determined by stan-
`dard PCR and hybridization methods.
`Statistical analysis. Statistical analysis was performed
`using the STATISTICA for Macintosh package (Stat-
`Soft, Tulsa, Oklahoma, USA) as indicated. Unless oth-
`erwise indicated, results are given as mean ± SEM.
`
`Results
`The in vitro proliferative response of PBMCs to GA decreases
`upon in vivo administration of GA. PBMCs were isolated
`from 7 patients with RR MS before and at various
`times after subcutaneous administration of GA. At
`each time point tested, primary and secondary in vitro
`proliferation and cytokine assays in the presence and
`absence of GA were performed. We found that before
`treatment, there was a significant proliferative
`
`Figure 2
`GA-specific secretion of cytokines is polarized toward a Th2 response after daily injections of GA. The GA-specific secretion of the cytokines
`IL-5 and IFN-g was measured in T-cell lines by 2 methods: ELISPOT (a) and ELISA (b) assays. Each symbol represents the difference of spots
`counted or D pg/mL measured in split-well assays between the GA (20 m g/mL) condition and the no-antigen control. The limits of detec-
`tion were 1 spot and 10 pg/mL, respectively. Numbers represent the percentage of T-cell lines in each quadrant with a minimum difference
`in spots of twice the SD of the negative controls for IL-5 and IFN-g , respectively.
`
`970
`
`The Journal of Clinical Investigation | April 2000 | Volume 105 | Number 7
`
`Page 4 of 10
`
`YEDA EXHIBIT NO. 2069
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`

`Figure 3
`IL-13 secretion is increased
`after daily injections of GA.
`Primary T-cell lines were set
`up in 10 identical wells each in
`the presence of no antigen or
`with 1.0, 10, and 100 m g/mL
`GA and cultured as described
`in Methods. Identical wells
`were pooled on day 11, and
`30,000 T cells each were res-
`timulated with no antigen or
`1.0, 10, and 100 m g/mL GA
`pulsed on 100,000 autolo-
`gous APCs. Cytokines were
`measured in supernatants by
`ELISA after 48 hours as
`described, and proliferation
`was measured by [3H]thymi-
`dine incorporation. Asterisks
`point at values that were
`above the upper limit of
`detection of the IL-13 assay of
`10,000 pg/mL.
`
`response as measured by [3H]thymidine incorporation
`to GA in all 7 patients, with an average stimulation
`index (SI) in vitro of 24.8 ± 1.1; the average D cpm was
`37,241 ± 1,766 cpm. Additionally, all of a total of 170
`independently derived T-cell lines stimulated in pri-
`mary in vitro culture with GA proliferated in response
`to the antigen (data not shown). After treatment with
`GA 20 mg subcutaneously daily for 3, 6 and 12
`months, the proliferative response as measured by SI
`and D
`cpm significantly decreased (Figure 1a) (P <
`0.001), although, as expected, individual patients var-
`ied in their response to GA (Figure 1b).
`In vitro–generated GA-reactive T-cell lines deviate toward a
`Th2-cytokine profile upon treatment with GA. Having
`demonstrated that the proliferative response to GA
`changed after in vivo subcutaneous administration of
`GA, we next examined whether the cytokine profile also
`changed. The cytokine response was measured for the
`prototypic Th1 and Th2 cytokines IFN-g and IL-5 by
`ELISPOT and sandwich ELISA in a total of 590 T-cell
`lines generated before and at various times after GA
`injection in all 7 patients. As shown in Figure 2, a and
`b, compared with the values detected before treatment,
`the average IFN-g secretion to GA measured by either
`ELISA or ELISPOT was significantly decreased (P <
`0.001 by Tukey’s honest statistical difference test) after
`treatment for 3, 6, and 12 months, except for the meas-
`urement by ELISPOT at 3 months, which did not reach
`significance, and the ELISA measurement at 6 months,
`which only reached a significance level of P < 0.01. The
`GA-dependent IFN-g
`secretion as determined by
`
`ELISPOT as D
`spots between the cells tested with anti-
`gen and the no-antigen control was 104 ± 10 before
`treatment, 132 ± 18 at 3 months, 28 ± 4 at 6 months,
`and 18 ± 3 at 12 months. IFN-g secretion measured by
`sandwich ELISA in D pg/mL was 1,405 ± 150 before
`treatment, 222 ± 38 at 3 months, 797 ± 185 at 6
`months, and 5.9 ± 46 at 12 months. When patients
`were analyzed individually, 5 of the 7 patients had sta-
`tistically significant decreases in IFN-g secretion (P <
`0.001) by ELISPOT or sandwich ELISA (data not
`shown). The levels of IFN-g secretion were correlated
`with the decreased proliferative capacity in these
`patients (r2 of > 0.8 for ELISA values in all patients test-
`ed, measurement by ELISPOT correlated less well with
`r2> 0.5 in 4 patients). This is in accordance with previ-
`ous observations that the proliferative and IFN-g
`responses are correlated (25).
`GA-specific IL-5 secretion was, on average, not sta-
`tistically significantly altered during treatment with
`GA. By ELISPOT assay, the average IL-5 secretion in D
`spots was 10 ± 3 before, 10 ± 2 at 3 months, 11 ± 2 at 6
`months, and 6 ± 1 at 12 months after the initiation of
`treatment. When measured by sandwich ELISA as D
`pg/mL, IL-5 secretion was 1,484 ± 266 before treat-
`ment; 1,940 ± 554 at 3 months; 1,738 ± 343 at 6
`months; and 1,146 ± 303 at 12 months after the initi-
`ation of treatment. One patient had a sustained sta-
`tistically significant decrease in IL-5 (P < 0.001), and 1
`had a sustained statistically significant increase (P <
`0.001) in IL-5 secretion as measured by ELISPOT dur-
`ing treatment.
`
`The Journal of Clinical Investigation | April 2000 | Volume 105 | Number 7
`
`971
`
`Page 5 of 10
`
`YEDA EXHIBIT NO. 2069
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`

`Figure 4
`Cross-reactivity of GA-reactive T-cell lines is increased after daily injections of GA. Percentages of the GA-induced T-cell lines cross-react-
`ing to each APL tested at each time point are shown for the 7 patients encoded by gray scale. Proliferative IFN-g and IL-5 responses were
`examined for all T-cell lines and are represented separately in the top, middle, and lower third. A minimum SI of 2 and a difference of
`2 SD over the background was required for classification as a cross-reactive T-cell line.
`
`To examine further the cytokine pattern of PBMCs
`from patients before and after treatment with GA, T-cell
`lines from all subjects were grouped into Th0, Th1, and
`Th2 subsets, based on their cytokine profile. T-cell lines
`were considered positive for a cytokine when the differ-
`ence of the GA condition was increased at least 2-fold
`over the SD of the no-antigen controls. Thus, values
`over background considered positive were 19 spots for
`IFN-g and 10 spots for the IL-5 ELISPOT assay, and 138
`pg/mL for IFN-g and 485 pg/mL for the IL-5 ELISA
`assays. When measured by ELISPOT before treatment,
`46% of all T cells evaluated were characterized as Th1
`(Figure 2a). During the course of treatment, there was
`an increased proportion at 3 months of Th0-type T-cell
`lines and at 6 and 12 months of Th2-type T-cell lines. In
`parallel with the decreased proliferative response, an
`increasing proportion of T-cell lines that did not secrete
`either IFN-g or IL-5 in response to GA was seen with
`treatment. Measurement by ELISA appears to be slight-
`ly more sensitive in this assay in which 39% of T-cell
`lines secreted Th0 cytokines and 41% secreted Th1
`cytokines before treatment (Figure 2b). A similar shift
`
`toward a Th2 response as with the ELISPOT assay with
`a decrease of Th0 and a simultaneous increase of Th2-
`type T-cell lines at 3, 6, and 12 months of treatment was
`seen. When classifications of T-cell lines into Th0, Th1,
`and Th2 were performed under less-stringent condi-
`tions (minimum difference of 20 and 10 ELISPOTS for
`IFN-g and IL-5, respectively, or 200 pg/mL in ELISA
`assays), a cytokine shift toward Th0/Th2 could also be
`confirmed (data not shown).
`As the magnitude of the IL-5 response was uniform-
`ly low, especially as measured by ELISPOT, it was
`important to reconfirm the apparent Th2 deviation
`with GA treatment by a means other than measuring
`IL-5. Toward this end, 3 new patients with RR MS were
`recruited to the study, and serial measurements of IFN-
`g and IL-13 secretion were made before and after the
`initiation of daily subcutaneous GA injections. IL-13
`was chosen as the candidate Th2 cytokine because
`there are no IL-13 receptors on T cells to consume the
`secreted cytokine. Primary in vitro T-cell lines generat-
`ed in the presence of no antigen and of 1.0, 10, and 100
`m g/mL GA were examined at 30,000 PBMCs per well
`
`972
`
`The Journal of Clinical Investigation | April 2000 | Volume 105 | Number 7
`
`Page 6 of 10
`
`YEDA EXHIBIT NO. 2069
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`

`each with the 4 antigen concentrations. Thus, equal
`numbers of cells were tested in the secondary stimula-
`tion, whereas in the previous assay with equal aliquots
`of primary cell lines, the number of cells tested
`decreased, on average, with treatment owing to lower
`expansion in the primary cultures. A marked increase
`in IL-13 secretion in 2 of 3 patients after 3 months of
`therapy was observed. IFN-g secretion and the prolifer-
`ative response were stable or decreased when compared
`with the pretreatment values (Figure 3).
`Cross-reactivity of GA-reactive T-cell lines is increased upon
`treatment with GA. Cross-reactivity of GA-induced T-cell
`lines to combinatorial peptide libraries derived from
`the immunodominant MBP 84-102 peptide and a com-
`pletely randomized 13mer library were performed to
`determine degeneracy of GA-specific T cells (Figure 4).
`Before treatment with GA, there was minimal cross-
`reactivity in either the proliferative or the cytokine
`responses. There was only 1 instance when antigen
`cross-reactivity was observed, in the proliferative
`response to the 93R90X combinatorial peptide library
`in patient 7. In striking contrast, 6 of the 7 patients
`demonstrated an increased number of cross-reactive T-
`
`cell lines after therapy to the combinatorial peptide
`libraries. However, no dominantly cross-reactive APL
`emerged from this analysis, consistent with the degen-
`erate immune responses we observed.
`In vitro T-cell reactivity to the immunodominant epitope
`MBP 84-102 is not significantly altered during GA therapy.
`In contrast to the results from primary in vitro cul-
`tures with GA, which is well approximated by a nor-
`mal distribution of responses owing to the high pre-
`cursor frequency of responsive T cells, the reactivity
`of primary T-cell lines to MBP 84-102 was low and as
`such not normally distributed. Therefore, nonpara-
`metric statistics were used for analysis. No signifi-
`cant change over time of treatment was seen for the
`MBP 84-102–specific proliferative responses (Figure
`5a) by Mann-Whitney U tests. Further analysis
`included percentage of MBP 84-102–specific T-cell
`lines, which were not significantly changed over the
`course of treatment.
`Analysis of cytokine secretion in response to MBP 84-
`102 by Mann-Whitney U test also did not reveal any sig-
`nificant differences during the course of treatment with
`one exception: IL-5 measured by ELISPOT at 12
`
`Figure 5
`(a). The proliferative response to MBP 84-102 is not altered after daily injections of GA. A total of 900 T-cell lines were generated in response to MBP
`84-102. The proliferative response in D cpm is given for each of 30 or 40 lines tested at each time point (circles, D cpm on left axes). On the right axes,
`the percentage of positive lines determined by a minimum 2.5-fold increase over background and a minimum difference of 1,500 cpm is shown (line).
`The mean background was 4,662 ± 138. (b). No changes occur in the cytokine response to MBP 84-102 after daily injections of GA. Lines having a
`minimum difference to the negative control of 2 SD, i.e., 70 spots for IFN-g and 19 spots for IL-5, were considered positive for the respective cytokine.
`
`The Journal of Clinical Investigation | April 2000 | Volume 105 | Number 7
`
`973
`
`Page 7 of 10
`
`YEDA EXHIBIT NO. 2069
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`

`months tested significantly decreased (P < 0.01) com-
`pared with pretreatment values. When the cytokines
`were analyzed for each patient individually, no signifi-
`cant change in the frequency of Th0-, Th1-, and Th2-
`cytokine–secreting lines was seen (Figure 5b), regardless
`of the stringency of the criteria used for classification.
`No cross-reactivity was seen between MBP 84-
`102–induced T cell lines and GA in vitro when judged
`by these criteria.
`GA treatment does not change tetanus toxoid–specific in vitro
`T-cell responses. To examine the effects of GA treatment
`on the primary in vitro T-cell response to an unrelated
`recall antigen, PBMCs from 6 patients with RR MS who
`were undergoing GA therapy were analyzed with
`tetanus toxoid. A dose-dependent proliferative response
`to 0.3 m g/mL, 3 m g/mL, and 30 m g/mL of tetanus toxoid
`as well as to 0.1 m g/mL and 1 m g/mL of the PHA control
`was observed at all time points. Compared with the pre-
`treatment values, no significant differences in the
`tetanus toxoid–specific proliferative responses were seen
`during treatment with GA as determined by the Stu-
`dent’s t test (data not shown). Thus, as with MBP 84-
`102, subcutaneous treatment with GA did not alter
`immune responses to a common recall antigen.
`
`Discussion
`We examined the effect of daily subcutaneous injec-
`tions of GA on antigen-specific T-cell responses in
`patients with MS. GA appears to function as a univer-
`sal antigen, inducing primary in vitro proliferation of
`naive T-cell populations both in patients with MS and
`in normal healthy controls. Daily subcutaneous injec-
`tions of GA caused a striking loss of in vitro respon-
`siveness to the GA that was accompanied by immune
`deviation to a more Th2 type of response. The surviv-
`ing GA-reactive T cells exhibited a greater degree of
`degeneracy as measured by cross-reactive responses to
`combinatorial peptide libraries. GA is, to our knowl-
`edge, the first agent effective in the treatment of an
`autoimmune disease that appears to alter immune
`function by engagement of the T-cell receptor and may
`be useful in a variety of autoimmune disorders in
`which immune deviation to a Th2 type of response
`may be desirable.
`Perhaps the most striking observation in these inves-
`tigations was the ability of GA to induce the prolifera-
`tion of non-GA–primed T-cell populations, which then
`decreased with treatment. This was not a nonspecific
`mitogenic response, as MHC DR-restricted T-cell
`clones could be generated with in vitro antigen culture
`(P.W. Duda, and D.A. Hafler, manuscript in prepara-
`tion). GA has been shown to directly bind different DR
`molecules without antigen processing, allowing many
`potential interactions with T-cell receptors (14).
`Although it had been thought that the recognition by
`the TCR of an MHC/peptide complex was highly spe-
`cific, it has recently become clear that there can be
`extensive degeneracy in TCR recognition of antigen in
`the trimolecular complex (26, 27). Moreover, we recent-
`
`ly demonstrated at a functional level that a TCR that
`may appear to be highly specific for 1 peptide/MHC
`complex may become significantly more degenerate in
`its recognition of antigen with subtle changes of amino
`acid side chains, particularl

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket